Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 15556, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38969656

RESUMO

Previously, we reported successful cellular expansion of a murine colorectal carcinoma cell line (CT-26) using a three-dimensional (3D) engineered extracellular matrix (EECM) fibrillar scaffold structure. CCL-247 were grown over a limited time period of 8 days on 3D EECM or tissue culture polystyrene (TCPS). Cells were then assayed for growth, electroporation efficiency and Vigil manufacturing release criteria. Using EECM scaffolds, we report an expansion of CCL-247 (HCT116), a colorectal carcinoma cell line, from a starting concentration of 2.45 × 105 cells to 1.9 × 106 cells per scaffold. Following expansion, 3D EECM-derived cells were assessed based on clinical release criteria of the Vigil manufacturing process utilized for Phase IIb trial operation with the FDA. 3D EECM-derived cells passed all Vigil manufacturing release criteria including cytokine expression. Here, we demonstrate successful Vigil product manufacture achieving the specifications necessary for the clinical trial product release of Vigil treatment. Our results confirm that 3D EECM can be utilized for the expansion of human cancer cell CCL-247, justifying further clinical development involving human tissue sample manufacturing including core needle biopsy and minimal ascites samples.


Assuntos
Matriz Extracelular , Imunoterapia , Alicerces Teciduais , Humanos , Alicerces Teciduais/química , Imunoterapia/métodos , Engenharia Tecidual/métodos , Células HCT116 , Neoplasias Colorretais/patologia , Animais , Camundongos , Proliferação de Células , Linhagem Celular Tumoral , Técnicas de Cultura de Células em Três Dimensões/métodos
2.
Vet Res Commun ; 39(4): 229-36, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26521056

RESUMO

Neonatal calf colibacillosis caused by enterotoxigenic Escherichia coli (ETEC) is an economically significant problem in most parts of the world. The most common ETEC found in calves express the F5 (K99) fimbriae, which are necessary for the attachment of the bacteria to the ganglioside receptors on enterocytes. It is known that prevention of ETEC F5(+) adhesion to its ganglioside receptors with specific antibodies protects calves from colibacillosis. Previously we have described the development and characterization of a mouse recombinant antibody fragment (moRAb) that prevents F5 fimbrial protein induced agglutination of horse red blood cells (HRBC), which exhibit the same gangloside receptor for F5 fimbriae. Here we demonstrate that this recombinant antibody fragment inhibits in vitro the attachment of ETEC F5(+) bacteria to HRBC as well as isolated calf enterocytes, and in vivo it decreases fluid accumulation in intestinal loops of calves. Thus, correct oral administration of this anti-F5 moRAb may serve as an immunoprophylactic for cost effective control of colibacillosis in calves.


Assuntos
Anticorpos Antibacterianos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Doenças dos Bovinos/prevenção & controle , Enterócitos/efeitos dos fármacos , Infecções por Escherichia coli/veterinária , Animais , Animais Recém-Nascidos , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/uso terapêutico , Bovinos , Doenças dos Bovinos/tratamento farmacológico , Doenças dos Bovinos/patologia , Enterotoxinas/toxicidade , Eritrócitos/efeitos dos fármacos , Escherichia coli , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/patologia , Infecções por Escherichia coli/prevenção & controle , Proteínas de Escherichia coli/imunologia , Fímbrias Bacterianas/patologia , Cavalos , Íleo/patologia , Masculino , Proteínas Recombinantes/imunologia
3.
Methods Mol Biol ; 942: 259-78, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23027056

RESUMO

The discovery of RNA interference (RNAi) engendered great excitement and raised expectations regarding its potential applications in biomedical research and clinical usage. Over the ensuing years, expanded understanding of RNAi and preliminary results from early clinical trials tempered enthusiasm with realistic appraisal resulting in cautious optimism and a better understanding of necessary research and clinical directions. As a result, data from more recent trials are beginning to show encouraging positive clinical outcomes. The capability of delivering a pharmacologically effective dose to the target site while avoiding adverse host reactions still remains a challenge although the delivery technology continues to improve. We have developed a novel vector-driven bifunctional short hairpin RNA (bi-shRNA) technology that harnesses both cleavage-dependent and cleavage-independent RISC loading pathways to enhance knockdown potency. Consequent advantages provided by the bi-shRNA include a lower effective systemic dose than comparator siRNA/shRNA to minimize the potential for off-target side effects, due to its ability to induce both a rapid (inhibition of protein translation) and delayed (mRNA cleavage and degradation) targeting effect depending on protein and mRNA kinetics, and a longer duration of effectiveness for clinical applications. Here, we provide an overview of key molecular methods for the design, construction, quality control, and application of bi-shRNA that we believe will be useful for others interested in utilizing this technology.


Assuntos
Engenharia Genética/métodos , RNA Interferente Pequeno/genética , Sequência de Bases , Colesterol/metabolismo , DNA/genética , DNA/metabolismo , Ácidos Graxos Monoinsaturados/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Sequências Repetidas Invertidas , Compostos de Amônio Quaternário/metabolismo , Clivagem do RNA , Interferência de RNA , Transfecção
4.
Mol Ther ; 20(3): 679-86, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22186789

RESUMO

We performed a phase I trial of FANG vaccine, an autologous tumor-based product incorporating a plasmid encoding granulocyte-macrophage colony-stimulating factor (GMCSF) and a novel bifunctional short hairpin RNAi (bi-shRNAi) targeting furin convertase, thereby downregulating endogenous immunosuppressive transforming growth factors (TGF) ß1 and ß2. Patients with advanced cancer received up to 12 monthly intradermal injections of FANG vaccine (1 × 10(7) or 2.5 × 10(7) cells/ml injection). GMCSF, TGFß1, TGFß2, and furin proteins were quantified by enzyme-linked immunosorbent assay (ELISA). Safety and response were monitored. Vaccine manufacturing was successful in 42 of 46 patients of whom 27 received ≥1 vaccine. There were no treatment-related serious adverse events. Most common grade 1, 2 adverse events included local induration (n = 14) and local erythema (n = 11) at injection site. Post-transfection mean product expression GMCSF increased from 7.3 to 1,108 pg/10(6) cells/ml. Mean TGFß1 and ß2 effective target knockdown was 93.5 and 92.5% from baseline, respectively. Positive enzyme-linked immunospot (ELISPOT) response at month 4 was demonstrated in 9 of 18 patients serially assessed and correlated with survival duration from time of treatment (P = 0.025). Neither dose-adverse event nor dose-response relationship was noted. In conclusion, FANG vaccine was safe and elicited an immune response correlating with prolonged survival. Phase II assessment is justified.


Assuntos
Vacinas Anticâncer/uso terapêutico , Furina/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Neoplasias/terapia , RNA Interferente Pequeno/uso terapêutico , Adulto , Idoso , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Feminino , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interferon gama/biossíntese , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias/imunologia , Neoplasias/mortalidade , Neoplasias/patologia , Análise de Sobrevida , Transgenes , Resultado do Tratamento
5.
DNA Cell Biol ; 30(9): 715-26, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21612405

RESUMO

Bifunctional small hairpin RNAs (bi-shRNAs) are functional miRNA/siRNA composites that are optimized for posttranscriptional gene silencing through concurrent mRNA cleavage-dependent and -independent mechanisms (Rao et al., 2010 ). We have generated a novel bi-shRNA using the miR30 scaffold that is highly effective for knockdown of human stathmin (STMN1) mRNA. STMN1 overexpression well documented in human solid cancers correlates with their poor prognosis. Transfection with the bi-shSTMN1-encoding expression plasmid (pbi-shSTMN1) markedly reduced CCL-247 human colorectal cancer and SK-Mel-28 melanoma cell growth in vitro (Rao et al., 2010 ). We now examine in vivo the antitumor efficacy of this RNA interference-based approach with human tumor xenografted athymic mice. A single intratumoral (IT) injection of pbi-shSTMN1 (8 µg) reduced CCL-247 tumor xenograft growth by 44% at 7 days when delivered as a 1,2-dioleoyl-3-trimethyl-ammoniopropane:cholesterol liposomal complex. Extended growth reductions (57% at day 15; p < 0.05) were achieved with three daily treatments of the same construct. STMN1 protein reduction was confirmed by immunoblot analysis. IT treatments with pbi-shSTMN1 similarly inhibited the growth of tumorgrafts derived from low-passage primary melanoma (≥70% reduction for 2 weeks) and abrogated osteosarcoma tumorgraft growth, with the mature bi-shRNA effector molecule detectable for up to 16 days after last injection. Antitumor efficacy was evident for up to 25 days posttreatment in the melanoma tumorgraft model. The maximum tolerated dose by IT injection of >92 µg (Human equivalent dose [HED] of >0.3 mg/kg) in CCL-247 tumor xenograft-bearing athymic mice was ∼10-fold higher than the extrapolated IC(50) of 9 µg (HED of 0.03 mg/kg). Healthy, immunocompetent rats were used as biorelevant models for systemic safety assessments. The observed maximum tolerated dose of <100 µg for intravenously injected pbi-shSTMN1 (mouse equivalent of <26.5 µg; HED of <0.09 mg/kg) confirmed systemic safety of the therapeutic dose, hence supporting early-phase assessments of clinical safety and preliminary efficacy.


Assuntos
Neoplasias Colorretais/terapia , Técnicas de Silenciamento de Genes/métodos , Terapia Genética/métodos , Melanoma/terapia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Estatmina/metabolismo , Animais , Primers do DNA/genética , Feminino , Humanos , Immunoblotting , Masculino , Dose Máxima Tolerável , Camundongos , Camundongos Nus , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Hum Gene Ther ; 22(11): 1331-41, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21517694

RESUMO

Hereditary inclusion body myopathy (HIBM) is an autosomal recessive adult-onset myopathy due to mutations in the GNE (UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase) gene. Affected patients have no therapeutic options. We have previously demonstrated in preclinical testing the ability to safely correct GNE gene function through liposomal delivery of the wild-type GNE gene. Results were verified in a single patient treated by intravenous infusion of GNE gene lipoplex. A single patient (patient 001) with severe HIBM treated with a compassionate investigational new drug received seven doses of GNE gene lipoplex via intravenous infusion at the following doses: 0.4, 0.4, 1.0, 4.0, 5.0, 6.0, and 7.0 mg of DNA. GNE transgene expression, downstream induction of sialic acid, safety, and muscle function were evaluated. Transient low-grade fever, myalgia, tachycardia, transaminase elevation, hyponatremia, and hypotension were observed after infusion of each dose of GNE gene lipoplex. Quadriceps muscle expression of the delivered GNE, plasmid, and RNA was observed 24 hr after the 5.0-mg dose and at significantly greater levels 72 hr after the 7.0-mg infusion in comparison with expression in quadriceps muscle immediately before infusion. Sialic acid-related proteins were increased and stabilization in the decline of muscle strength was observed. We conclude that clinical safety and activity have been demonstrated with intravenous infusion of GNE gene lipoplex. Further assessment will involve a phase I trial of intravenous administration of GNE gene lipoplex in individuals with less advanced HIBM with more muscle function.


Assuntos
Complexos Multienzimáticos/genética , Miosite de Corpos de Inclusão/terapia , Adulto , Feminino , Terapia Genética , Vetores Genéticos , Humanos , Infusões Intravenosas , Lipossomos , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Miosite de Corpos de Inclusão/genética , RNA/metabolismo
7.
J Gene Med ; 12(5): 403-12, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20440751

RESUMO

BACKGROUND: Hereditary inclusion body myopathy (HIBM) is an autosomal recessive adult onset myopathy. It is characterized by mutations of the GNE (UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase) gene. Afflicted patients have no therapeutic options. In preclinical testing, we have previously demonstrated the ability to correct GNE gene function and the safety of delivery of wild type GNE gene using a liposomal delivery vehicle. METHODS: A single patient (subject #001) with severe HIBM treated by compassionate investigational new drug received four doses of GNE gene Lipoplex via intramuscular injection. GNE transgene expression, downstream induction of sialic acid, safety and muscle function were evaluated. RESULTS: Significant durable improvement in locoregional skeletal muscle function was observed in the injected left extensor carpi radialis longus of #001 in correlation with GNE transgene upregulation and local induction of sialic acid. Other than transient low grade fever and pain at the injection site, no significant toxicity was observed. CONCLUSIONS: Proof of principle for manufacturing of 'clinical grade' GNE gene Lipoplex, clinical safety and activity are demonstrated with GNE gene Lipoplex. Further assessment will involve intravenous administration and subsequent phase I trial involving additional but less severely afflicted HIBM patients.


Assuntos
Terapia Genética , Lipossomos/metabolismo , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/uso terapêutico , Miosite de Corpos de Inclusão/genética , Miosite de Corpos de Inclusão/terapia , Adolescente , Adulto , Biópsia , Feminino , Terapia Genética/efeitos adversos , Humanos , Injeções Intramusculares , Força Muscular , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Miosite de Corpos de Inclusão/fisiopatologia , Ácido N-Acetilneuramínico/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Adulto Jovem
8.
Gene Regul Syst Bio ; 3: 89-101, 2009 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-19838336

RESUMO

Hereditary inclusion body myopathy-2 (HIBM2) is an adult-onset, muscular disease caused by mutations in the GNE gene. HIBM2-associated GNE mutations causing hyposialyation have been proposed to contribute to reduced muscle function in patients with HIBM2, though the exact cause of this disease is unknown. In the current studies we examined pre-clinical in vivo toxicity, and expression of the plasmid-based, CMV driven wild-type GNE plasmid vector. The plasmid vector was injected intramuscularly (IM) or systemically (IV) into BALB/c mice, following encapsulation in a cationic liposome (DOTAP:Cholesterol). Single IM injections of the GNE-lipoplex at 40 microg did not produce overt toxicity or deaths, indicating that the no observable adverse effect level (NOAEL) dose for IM injection was >or=40 microg. Single intravenous (IV) infusion of GNE-lipoplex was lethal in 33% of animals at 100 microg dose, with a small proportion of animals in the 40 microg cohort demonstrating transient toxicity. Thus the NOAEL dose by the IV route was greater than 10 microg and less than or equal to 40 microg. Real-time RT-qPCR analysis demonstrated recombinant human GNE mRNA expression in 100% of muscle tissues that received IM injection of 40 microg GNE-lipoplex, at 2 weeks. These results indicate that GNE-lipoplex gene transfer is safe and can produce durable transgene expression in treated muscles. Our findings support future exploration of the clinical efficacy of GNE-lipoplex for experimental gene therapy of HIBM2.

9.
Curr Gene Ther ; 9(1): 45-60, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19275571

RESUMO

Tremendous strides have been made in proteogenomics and RNA interference technologies. Hence "personalized" cancer gene therapy has become a foreseeable rather than a predictable reality. Currently, the lack of an optimized, systemic gene delivery vehicle remains a key limiting factor for developing effective treatment applications. Since their introduction by Felgner in 1987, cationic lipids have been an attractive consideration for gene delivery, in view of their biocompatibility, biodegradability, low toxicity, and low immunogenicity. Successful in vivo transgene expression by cationic lipid- or cationic polymer-based delivery depends critically on a long circulating half life (>48 h), a definable systemic biodistribution with target-specific cancer localization, and efficient cell entry and internalization. Ideally, the agent should have a hydrophobic, stabilized core that ensures integrity of the therapeutic entity in vivo, a biocompatible, neutrally charged shell (zeta potential of approximately +/-10 mv) for enhanced, "stealth" circulation, and a suitable size (approximately 50-200 nm in diameter) for access into the tumor neovasculature and reduced reticuloendothelial system (RES) uptake. "Smart" receptor-targeting moieties can redirect intracellular trafficking. Additional engineered features have also been incorporated to minimize lysosomal degradation (membrane fusogenic lipids or proton sponge), promote endosomal escape into cytoplasm (cell penetrating peptides, triblock copolymer construction), and enhance nuclear entry and activate the endogenous transcriptional machinery (inclusion of a nuclear localization signal). Improvements in each of these respective areas of study have converged to yield promising in vivo results.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos , Lipídeos/administração & dosagem , Neoplasias/terapia , Animais , Humanos , Neoplasias/genética
10.
Gene Regul Syst Bio ; 3: 181-90, 2009 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20054407

RESUMO

Hereditary inclusion body myopathy type 2 (HIBM2) is a myopathy characterized by progressive muscle weakness with early adult onset. The disease is the result of a recessive mutation in the Glucosamine (UDP-N-acetyl)-2-epimerase/N-acetylmannosamine kinase gene (GNE), which results in reduced enzyme function and sialic acid levels. A majority of individuals with HIBM2 are from Iranian-Jewish or Japanese decent, but isolated cases have been identified world wide. This article reviews the diagnostic criteria for HIBM2. Current research with a highlight on the biology of the disease and the role of GNE in the sialic acid pathway are assessed. Finally, therapeutic investigations and animal models are discussed with a focus on future studies to better understand the pathology of Hereditary Inclusion Body Myopathy and move therapeutic agents towards clinical trials.

11.
Glycoconj J ; 26(4): 415-22, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18815882

RESUMO

The bifunctional enzyme UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) is the key enzyme for the biosynthesis of sialic acids, terminal components of glycoconjugates associated with a variety of physiological and pathological processes. Different protein isoforms of human and mouse GNE, deriving from splice variants, were predicted recently: GNE1 represents the GNE protein described in several studies before, GNE2 and GNE3 are proteins with extended and deleted N-termini, respectively. hGNE2, recombinantly expressed in insect and mamalian cells, displayed selective reduction of UDP-GlcNAc 2-epimerase activity by the loss of its tetrameric state, which is essential for full enzyme activity. hGNE3, which had to be expressed in Escherichia coli, only possessed kinase activity, whereas mGNE1 and mGNE2 showed no significant differences. Our data therefore suggest a role of GNE1 in basic supply of cells with sialic acids, whereas GNE2 and GNE3 may have a function in fine-tuning of the sialic acid pathway.


Assuntos
Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Camundongos , Estrutura Quaternária de Proteína , Proteínas Recombinantes/metabolismo
12.
J Biol Chem ; 283(46): 32056-65, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18805794

RESUMO

Growth factor independent-1 (Gfi1) is a zinc finger protein with a SNAG-transcriptional repressor domain. Ajuba is a LIM domain protein that shuttles between the cytoplasm and the nucleus. Ajuba functions as a co-repressor for synthetic Gfi1 SNAG-repressor domain-containing constructs, but a role for Ajuba co-repression of the cognate DNA bound Gfi1 protein has not been defined. Co-immunoprecipitation of synthetic and endogenous proteins and co-elution with gel filtration suggest that an endogenous Ajuba.Gfi1.HDAC multiprotein complex is possible. Active histone deacetylase activity co-immunoprecipitates with Ajuba or Gfi1, and both proteins depend upon histone deacetylases for full transcriptional repression activity. Ajuba LIM domains directly bind to Gfi1, but the association is not SNAG domain-dependent. ChIP analysis and reciprocal knockdown experiments suggest that Ajuba selectively functions as a co-repressor for Gfi1 autoregulation. The data suggest that Ajuba is utilized as a corepressor selectively on Gfi1 target genes.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Histona Desacetilases/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Humanos , Proteínas com Domínio LIM , Ligação Proteica , Fatores de Transcrição/genética
13.
Gene Regul Syst Bio ; 2: 243-52, 2008 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-19787087

RESUMO

Hereditary Inclusion Body Myopathy (HIBM2) is a chronic progressive skeletal muscle wasting disorder which generally leads to complete disability before the age of 50 years. There is currently no effective therapeutic treatment for HIBM2. Development of this disease is related to expression in family members of an autosomal recessive mutation of the GNE gene, which encodes the bifunctional enzyme UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE/MNK). This is the rate limiting bifunctional enzyme that catalyzes the first 2 steps of sialic acid biosynthesis. Decreased sialic acid production, consequently leads to decreased sialyation of a variety of glycoproteins including the critical muscle protein alpha-dystroglycan (alpha-DG). This in turn severely cripples muscle function and leads to the onset of the syndrome. We hypothesize that replacing the mutated GNE gene with the wildtype gene may restore functional capacity of GNE/MNK and therefore production of sialic acid, allowing for improvement in muscle function and/or delay in rate of muscle deterioration. We have constructed three GNE gene/CMV promoter plasmids (encoding the wildtype, HIBM2, and Sialuria forms of GNE) and demonstrated enhanced GNE gene activity following delivery to GNE-deficient CHO-Lec3 cells. GNE/MNK enzyme function was significantly increased and subsequent induction of sialic acid production was demonstrated after transfection into Lec3 cells with the wild type or R266Q mutant GNE vector. These data form the foundation for future preclinical and clinical studies for GNE gene transfer to treat HIBM2 patients.

14.
DNA Cell Biol ; 26(5): 293-300, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17504025

RESUMO

MicroRNAs (miRNAs) are a recently discovered class of small (approximately 18-24 nt) nucleic acids that negatively regulate gene expression. This novel class of molecules modulates a wide array of growth and differentiation processes in human cancers. High throughput analyses, utilizing the solid phase, array platform, or liquid phase, bead-based hybridization have variously demonstrated that miRNA expression was commonly dysregulated in human cancer. miRNA expression profiling has shown promise in defining malignant status in retrospective studies. Considerable disagreement remains with respect to the miRNA signature for a specific cancer cell type, which appears to depend largely on the analytical platform. Nonetheless, various internally controlled studies have successfully identified the histotype of tumors of unknown origin according to miRNA expression profile. The evaluation of miRNAs expression may also be of prognostic value, as best exemplified by the correlation of let-7 and mir-155 levels with disease survival in nonsmall cell lung cancer.


Assuntos
MicroRNAs/genética , Neoplasias/diagnóstico , Neoplasias/genética , Animais , Biomarcadores/análise , Perfilação da Expressão Gênica , Humanos , MicroRNAs/análise , Valor Preditivo dos Testes , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...